The lymphoma-associated NPM-ALK oncogene elicits a p16INK4a/pRb-dependent tumor-suppressive pathway by Paola Martinelli, Paola Bonetti, Cristina Sironi,

Slides:



Advertisements
Similar presentations
Figure S1 A B Figure S1. SPATA2 is required for TNFα or zVAD.fmk induced necroptosis in L929 cells. (A) L929 cells were transfected with a pool of four.
Advertisements

Up-Regulation of Activating Transcription Factor-5 Suppresses SAP Expression to Activate T Cells in Hemophagocytic Syndrome Associated with Epstein-Barr.
by Elena A. Federzoni, Peter J. M. Valk, Bruce E
Myc sensitizes p53-deficient cancer cells to the DNA-damaging effects of the DNA methyltransferase inhibitor decitabine by Andreas Höglund, Lisa M. Nilsson,
The effect of human tissue factor pathway inhibitor-2 on the growth and metastasis of fibrosarcoma tumors in athymic mice by Hitendra Singh Chand, Xin.
Volume 30, Issue 3, Pages (May 2008)
Antiangiogenic antithrombin down-regulates the expression of the proangiogenic heparan sulfate proteoglycan, perlecan, in endothelial cells by Weiqing.
by Sang-Hyun Song, AeRi Kim, Tobias Ragoczy, M. A
PML Is a Direct p53 Target that Modulates p53 Effector Functions
The C/EBPδ tumor suppressor is silenced by hypermethylation in acute myeloid leukemia by Shuchi Agrawal, Wolf-Karsten Hofmann, Nicola Tidow, Mathias Ehrich,
The TCL1 oncoprotein inhibits activation-induced cell death by impairing PKCθ and ERK pathways by Gilles Despouy, Marjorie Joiner, Emilie Le Toriellec,
FOG-1 represses GATA-1-dependent FcϵRI β-chain transcription: transcriptional mechanism of mast-cell-specific gene expression in mice by Keiko Maeda, Chiharu.
CD74 induces TAp63 expression leading to B-cell survival
Multiple Tumor Suppressor Pathways Negatively Regulate Telomerase
Autocrine release of interleukin-9 promotes Jak3-dependent survival of ALK+ anaplastic large-cell lymphoma cells by Lin Qiu, Raymond Lai, Quan Lin, Esther.
Volume 12, Issue 1, Pages (January 2013)
PTF1α/p48 and cell proliferation
Modulation of K-Ras-Dependent Lung Tumorigenesis by MicroRNA-21
Hyaluronate-Enhanced Hematopoiesis: Two Different Receptors Trigger the Release of Interleukin-1β and Interleukin-6 From Bone Marrow Macrophages by Sophia.
PRDM1/BLIMP1 is commonly inactivated in anaplastic large T-cell lymphoma by Michela Boi, Andrea Rinaldi, Ivo Kwee, Paola Bonetti, Maria Todaro, Fabrizio.
Surface IgM stimulation induces MEK1/2-dependent MYC expression in chronic lymphocytic leukemia cells by Sergey Krysov, Samantha Dias, Alex Paterson, C.
Volume 4, Issue 5, Pages (November 2003)
Volume 43, Issue 4, Pages (August 2011)
Constitutive STAT6 activation in primary mediastinal large B-cell lymphoma by Chrystelle Guiter, Isabelle Dusanter-Fourt, Christiane Copie-Bergman, Marie-Laure.
NPM-ALK oncogenic kinase promotes cell-cycle progression through activation of JNK/cJun signaling in anaplastic large-cell lymphoma by Vasiliki Leventaki,
LPS induces CD40 gene expression through the activation of NF-κB and STAT-1α in macrophages and microglia by Hongwei Qin, Cynthia A. Wilson, Sun Jung Lee,
The interferon regulatory factor ICSBP/IRF-8 in combination with PU
E2F4 loss suppresses tumorigenesis in Rb mutant mice
by Dong Yun Lee, and Bill Sugden
Volume 8, Issue 2, Pages (August 2001)
Volume 138, Issue 5, Pages e2 (May 2010)
Volume 18, Issue 3, Pages (April 2005)
Level of EGFR inhibition determines cell death response in EGFR mutant GBM cells. Level of EGFR inhibition determines cell death response in EGFR mutant.
P16Ink4a Suppression of Lung Adenocarcinoma by Bmi-1 in the Presence of p38 Activation  Mi-Ok Lee, MSc, Hyeon-Jae Lee, MD, Mi-Ae Kim, MD, Eun-Kyung Kim,
Oliver I. Fregoso, Shipra Das, Martin Akerman, Adrian R. Krainer 
Tomoyasu Hattori, Lukasz Stawski, Sashidhar S
c-Jun Promotes whereas JunB Inhibits Epidermal Neoplasia
Volume 7, Issue 6, Pages (June 2005)
MiTF Regulates Cellular Response to Reactive Oxygen Species through Transcriptional Regulation of APE-1/Ref-1  Feng Liu, Yan Fu, Frank L. Meyskens  Journal.
Volume 142, Issue 7, Pages e2 (June 2012)
Volume 18, Issue 13, Pages (March 2017)
LATS2 promotes death of lumB cells.
Volume 12, Issue 2, Pages (August 2003)
Absence of Distinguishing Senescence Traits in Human Melanocytic Nevi
PRAK Is Essential for ras-Induced Senescence and Tumor Suppression
Glucose-Induced β-Catenin Acetylation Enhances Wnt Signaling in Cancer
Minchul Kim, Taekhoon Kim, Randy L. Johnson, Dae-Sik Lim  Cell Reports 
Volume 19, Issue 6, Pages (June 2011)
14-3-3σ Regulates Keratinocyte Proliferation and Differentiation by Modulating Yap1 Cellular Localization  Sumitha A.T. Sambandam, Ramesh B. Kasetti,
FOXO3a Is Activated in Response to Hypoxic Stress and Inhibits HIF1-Induced Apoptosis via Regulation of CITED2  Walbert J. Bakker, Isaac S. Harris, Tak.
Volume 54, Issue 5, Pages (June 2014)
Volume 7, Issue 1, Pages (January 2008)
Inhibition of PAX3 by TGF-β Modulates Melanocyte Viability
Barbara Marinari, Costanza Ballaro, Maranke I
Hung-Chun Chang, Leonard Guarente  Cell 
Transcriptional Repression of miR-34 Family Contributes to p63-Mediated Cell Cycle Progression in Epidermal Cells  Dario Antonini, Monia T. Russo, Laura.
Short Telomeres in ESCs Lead to Unstable Differentiation
Volume 17, Issue 4, Pages (April 2010)
Volume 24, Issue 12, Pages (September 2018)
Volume 25, Issue 11, Pages e6 (December 2018)
Transcriptional Regulation of AKT Activation by E2F
Volume 6, Issue 3, Pages (September 2004)
Oliver I. Fregoso, Shipra Das, Martin Akerman, Adrian R. Krainer 
Volume 14, Issue 1, Pages (January 2001)
PU.1 Expression Delineates Heterogeneity in Primary Th2 Cells
Volume 2, Issue 3, Pages (September 2012)
The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells.
p53β regulates cellular senescence.
TAMs upregulate DNMT1 in gastric cancer cells through the CCL5/CCR5/STAT3 pathway. TAMs upregulate DNMT1 in gastric cancer cells through the CCL5/CCR5/STAT3.
The tumor-suppressive functions of the human INK4A locus
Presentation transcript:

The lymphoma-associated NPM-ALK oncogene elicits a p16INK4a/pRb-dependent tumor-suppressive pathway by Paola Martinelli, Paola Bonetti, Cristina Sironi, Giancarlo Pruneri, Caterina Fumagalli, Paola Rafaniello Raviele, Sara Volorio, Stefano Pileri, Roberto Chiarle, Fiona Kate Elizabeth McDuff, Betsabeh Khoramian Tusi, Suzanne D. Turner, Giorgio Inghirami, Pier Giuseppe Pelicci, and Emanuela Colombo Blood Volume 117(24):6617-6626 June 16, 2011 ©2011 by American Society of Hematology

NPM-ALK inhibits p53 activity and it induces a senescent checkpoint in primary fibroblasts. NPM-ALK inhibits p53 activity and it induces a senescent checkpoint in primary fibroblasts. (A-F) WT MEFs were infected with control (Ctrl) and NPM-ALK (NA) or RASV12 expressing retroviruses. (A) Cells were analyzed (5 days after infection) by immunofluorescence (magnification ×600) and Western blot (B) with an anti-γH2AX antibody (representative micrographs and percentage of labeled cells are shown). (C) Growth curve of infected cells plated after infection and counted daily (cell numbers are expressed relative to day 1; *P < .05 relative to control cells infected with an empty vector). (D) β-galactosidase detection (representative micrographs [magnification ×600] and percentage of positive cells) and anti-BrdU staining (percentage of positive cells relative to control cells) performed 6 and 4 days after infection, respectively. (E) Western blotting of p53, ARF, p16INK4a, NPM-ALK, and tubulin expression, 4 days after infection. (F) Western blotting of pRb, NPM-ALK, cyclin A, and tubulin expression, 3 days after infection. (G) Luciferase assay in WT MEFs transduced with the pGL13 p53-reporter, the CMV-βGal vector, and increasing amounts of vectors expressing NPM-ALK or the K210R kinase-dead NPM-ALK mutant (or a control empty vector). Data are from 3 independent experiments and normalized to βGal. *P < .05 relative to control cells infected with an empty vector. (H) Same Luciferase assay as in panel G comparing a single dose (100 ng) of NPM-ALK– and RASV12-expressing vectors. Paola Martinelli et al. Blood 2011;117:6617-6626 ©2011 by American Society of Hematology

NPM-ALK induced senescence requires an intact p16INK4a/pRb pathway. NPM-ALK induced senescence requires an intact p16INK4a/pRb pathway. Growth curves of (A) p16INK4a−/− or (B) Rb1−/− MEFs infected with the indicated retroviruses. Values are expressed relative to day 1. In each panel, results are representative of at least 3 independent experiments. *P values are relative to control cells infected with empty vectors. (C) Methylcellulose-CFU of WT or p16INK4a−/− lin− cells infected with the indicated retroviruses (graph and representative images). Values are expressed relative to control infected cells. (D) Serial replating of WT and p16INK4a−/− lin− cells infected with the indicated retroviruses. CFUs are expressed relative to the first count for each sample. Results are representative of 3 experiments. *P < .05 relative to control cells infected with an empty vector. Paola Martinelli et al. Blood 2011;117:6617-6626 ©2011 by American Society of Hematology

NPM-ALK activates the p16INK4a pathway in vivo. NPM-ALK activates the p16INK4a pathway in vivo. (A) SA-β-gal staining of frozen sections from 1.5-month-old mice of the indicated genotypes (representative images [magnification ×400] and percentage of positive cells). (B) Percentage of Ki67-positive cells from thymocyte cell-suspensions of 1.5-month-old NPM-ALK transgenic or control littermates, as evaluated by FACS analysis. Results were obtained from 3 independent experiments, each performed using at least 2 mice of each genotype. (C) Survival curves of NPM-ALK transgenic mice of different genotypes for p16INK4a, as indicated. Median time to death is indicated in the inset. (D) Western blot analyses of p16INK4a, p53, and ARF expression in tumors derived from NPM-ALK transgenic mice of different p16INK4a genotypes, as indicated. Tubulin or vinculin was used as loading control. C+ indicates WT MEFs (positive control); C−, p16INK4a- or Tp53-null MEFs (negative controls). Paola Martinelli et al. Blood 2011;117:6617-6626 ©2011 by American Society of Hematology

Expression of p16INK4a, p53, and pRb in 19 tumor specimens from ALCL ALK+ patients by immunohistochemical analysis with specific antibodies. Expression of p16INK4a, p53, and pRb in 19 tumor specimens from ALCL ALK+ patients by immunohistochemical analysis with specific antibodies Paola Martinelli et al. Blood 2011;117:6617-6626 ©2011 by American Society of Hematology

p16INK4a reactivation limits growth of ALCL cell lines. p16INK4a reactivation limits growth of ALCL cell lines. (A) Western blotting analysis of pRb, p16INK4a, phosphorylated pRb (serine 807-811), NPM-ALK, and tubulin expression in 6 different ALCL cell lines (as indicated). (B) Left panel: Western blot analysis of NPM-ALK, p16INK4a, and tubulin expression at different times (as indicated) in SU-DHL-1 cells treated with doxocyclin to induce shRNA interference of NPM-ALK. Right panel: p16INK4a mRNA quantification by qPCR analysis with specific primers. Data were standardized with ribosomal RNA and normalized against control untreated cells. (C) Karpas299 and DEL cell lines were infected with control (Ctrl) and p16INK4a-expressing retroviruses. Left panel: at day 4, cells were collected and expression of p16INK4a, phosphoRb, and tubulin evaluated by Western blot. Right panels: infected cells were selected, plated, and counted daily. (D) Karpas299 and DEL cell lines were treated with 5-azacytidine (AZA) for 6 days. Left panel: at day 5, cells were collected and expression of p16INK4a, phosphoRb, and tubulin evaluated by Western blot. Right panels: cells were plated and counted daily. Paola Martinelli et al. Blood 2011;117:6617-6626 ©2011 by American Society of Hematology

NPM-ALK induces p16INK4a promoter de-methylation. NPM-ALK induces p16INK4a promoter de-methylation. (A) Top panel: chromatin immunoprecipitation analysis of p16INK4a promoter H3K27me3 levels in WT MEFs infected with the indicated retroviruses. Precipitated DNA was amplified by qPCR using primers located in the p16INK4a promoter region. Enrichments are presented as percentage of total input DNA. Bottom panel: Q-PCR analysis of p16INK4a mRNA levels in the same cells, expressed as fold increase with respect to the day 1 control vector. (B) Growth curves of WT MEFs infected with a control lentivirus or lentiviruses expressing JunB-specific shRNAs, in the presence or absence of NPM-ALK, as indicated. Values are expressed relative to day 1 for each sample. *P < .05 with respect to control cells infected with a control vector. (C) Western blotting of p16INK4a, JunB, and NPM-ALK expression in the same cell samples as in panel B, 4 days after infection. Tubulin was used as a loading control. p16INK4a protein levels were calculated by densimetric analysis and normalized to control (Ctrl.) and tubulin band intensities. (D) Western blotting of Jmjd3 and p16INK4a expression levels in WT MEFs infected with NPM-ALK or control viruses. p16INK4a protein levels were calculated as in panel C. (E) Growth curves of WT and Jmjd3 null MEFs infected as indicated. Values are expressed relative to day 1 for each sample. (F) Western blotting of p16INK4a and NPM-ALK expression levels in WT and Jmjd3-null MEFs in the same cell samples as in panel E, 4 days after infection. p16INK4a protein levels were calculated as in panel C. (G) Left panel: Western blot analysis of STAT3, p16INK4a, and tubulin expression at different times (as indicated) in TS cells treated with doxocyclin to induce shRNA interference targeting STAT3. Right panel: p16INK4a mRNA quantification by qPCR analysis with specific primers. Data were standardized with ribosomal RNA and normalized against control untreated cells. Paola Martinelli et al. Blood 2011;117:6617-6626 ©2011 by American Society of Hematology